EP2534149B1 - 6-amino-2-{[(1s)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8h-purin-8-one maleate - Google Patents

6-amino-2-{[(1s)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8h-purin-8-one maleate Download PDF

Info

Publication number
EP2534149B1
EP2534149B1 EP11709025.8A EP11709025A EP2534149B1 EP 2534149 B1 EP2534149 B1 EP 2534149B1 EP 11709025 A EP11709025 A EP 11709025A EP 2534149 B1 EP2534149 B1 EP 2534149B1
Authority
EP
European Patent Office
Prior art keywords
purin
methylbutyl
oxy
piperidinyl
dihydro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP11709025.8A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2534149A1 (en
Inventor
Robert Hermann Gibbon
Amanda Lucas
Stephen Andrew Hermitage
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline LLC
Original Assignee
GlaxoSmithKline LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by GlaxoSmithKline LLC filed Critical GlaxoSmithKline LLC
Priority to SI201130364T priority Critical patent/SI2534149T1/sl
Priority to MEP-2014-154A priority patent/ME01926B/me
Priority to PL11709025T priority patent/PL2534149T3/pl
Publication of EP2534149A1 publication Critical patent/EP2534149A1/en
Application granted granted Critical
Publication of EP2534149B1 publication Critical patent/EP2534149B1/en
Priority to CY20141101069T priority patent/CY1116091T1/el
Priority to HRP20141258AT priority patent/HRP20141258T1/hr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present invention relates to compounds, processes for their preparation, compositions containing them, to their use in the treatment of various disorders in particular allergic diseases and other inflammatory conditions for example allergic rhinitis and asthma, infectious diseases, cancer, and as vaccine adjuvants.
  • allergic diseases and other inflammatory conditions for example allergic rhinitis and asthma, infectious diseases, cancer, and as vaccine adjuvants.
  • Vertebrates are constantly threatened by the invasion of microorganisms and have evolved mechanisms of immune defence to eliminate infective pathogens.
  • this immune system comprises two branches; innate immunity and acquired immunity.
  • the first line of host defence is the innate immune system, which is mediated by macrophages and dendritic cells.
  • Acquired immunity involves the elimination of pathogens at the late stages of infection and also enables the generation of immunological memory. Acquired immunity is highly specific, due to the vast repertoire of lymphocytes with antigen-specific receptors that have undergone gene rearrangement.
  • the innate immune response was originally thought to be non-specific, but is now known to be able to discriminate between self and a variety of pathogens.
  • the innate immune system recognises microbes via a limited number of germline-encoded Pattern-Recognition Receptors (PRRs) which have a number of important characteristics.
  • PRRs Pattern-Recognition Receptors
  • TLRs Toll-like receptors
  • TLRs are a family of ten Pattern Recognition Receptors described in man. TLRs are expressed predominantly by innate immune cells where their role is to monitor the environment for signs of infection and, on activation, mobilise defence mechanisms aimed at the elimination of invading pathogens.
  • the early innate immune-responses triggered by TLRs limit the spread of infection, while the pro-inflammatory cytokines and chemokines that they induce lead to recruitment and activation of antigen presenting cells, B cells, and T cells.
  • the TLRs can modulate the nature of the adaptive immune-responses to give appropriate protection via dendritic cell-activation and cytokine release ( Akira S., et al, Nat. Immunol., 2001: 2, 675-680 ).
  • the profile of the response seen from different TLR agonists depends on the cell type activated.
  • TLR7 is a member of the subgroup of TLRs (TLRs 3, 7, 8, and 9), localised in the endosomal compartment of cells which have become specialised to detect non-self nucleic acids. TLR7 plays a key importance in anti-viral defence via the recognition of ssRNA ( Diebold S.S., et al, Science, 2004: 303, 1529-1531 ; and Lund J. M., et al, PNAS, 2004: 101, 5598-5603 ). TLR7 has a restricted expression-profile in man and is expressed predominantly by B cells and plasmacytoid dendritic cells (pDC), and to a lesser extent by monocytes.
  • pDC plasmacytoid dendritic cells
  • Plasmacytoid DCs are a unique population of lymphoid-derived dendritic cells (0.2-0.8% of Peripheral Blood Mononuclear Cells (PBMCs)) which are the primary type I interferon-producing cells secreting high levels of interferon-alpha (IFN ⁇ ) and interferon-beta (IFN ⁇ ) in response to viral infections ( Liu Y-J, Annu. Rev. Immunol., 2005: 23, 275-306 ).
  • PBMCs Peripheral Blood Mononuclear Cells
  • Allergic diseases are associated with a Th2-biased immune-response to allergens.
  • Th2 responses are associated with raised levels of IgE, which, via its effects on mast cells, promotes a hypersensitivity to allergens, resulting in the symptoms seen, for example, in allergic rhinitis.
  • IgE IgE
  • Th2/Th1 regulatory T cell response.
  • TLR7 ligands have been shown to reduce Th2 cytokine and enhance Th1 cytokine release in vitro and to ameliorate Th2-type inflammatory responses in allergic lung models in vivo ( Fili L., et al, J. All. Clin.
  • TLR7 ligands have the potential to rebalance the immune-response seen in allergic individuals and lead to disease modification.
  • Interferon was first described as a substance which could protect cells from viral infection ( Isaacs & Lindemann, J. Virus Interference. Proc. R. Soc. Lon. Ser. B. Biol. Sci. 1957: 147, 258-267 ).
  • the type I interferons are a family of related proteins encoded by genes on chromosome 9 and encoding at least 13 isoforms of interferon alpha (IFN ⁇ ) and one isoform of interferon beta (IFN ⁇ ).
  • Recombinant IFN ⁇ was the first approved biological therapeutic and has become an important therapy in viral infections and in cancer.
  • interferons are known to be potent modulators of the immune response, acting on cells of the immune system.
  • interferon combinations can be highly effective at reducing viral load and in some subjects in eliminating viral replication.
  • many patients fail to show a sustained viral response and in these patients viral load is not controlled.
  • therapy with injected interferon may be associated with a number of unwanted adverse effects which are shown to affect compliance ( Dudley T., et al, Gut., 2006: 55(9), 1362-3 ).
  • imiquimod demonstrated adjuvant activities either topically ( Adams S., et al, J. Immunol., 2008, 181:776-84 ; Johnston D., et al, Vaccine, 2006, 24:1958-65 ), or systemically ( Fransen F. et al, Infect. Immun., 2007, 75:5939-46 ).
  • Resiquimod and other related TLR7/8 agonists have also been shown to display adjuvant activity ( Ma R. et al, Biochem. Biophys. Res. Commun., 2007, 361:537-42 ; Wille-Reece U., et al, Proc. Natl. Acad. Sci. USA, 2005, 102:15190-4 ; Wille-Reece U., et al, US2006045885 A1 ).
  • pDCs plasmacytoid dendritic cells
  • TLR7 and TLR9 plasmacytoid dendritic cells
  • stimulation of these receptors with viral RNA or DNA respectively can induce expression of interferon alpha.
  • TLR7 agonists include imidazoquinoline compounds such as imiquimod and resiquimod, oxoadenine analogues and also nucleoside analogues such as loxoribine and 7-thia-8-oxoguanosine which have long been known to induce interferon alpha.
  • International Patent Application publication number WO 2008/114008 discloses 9-subsituted-8-oxoadenine compounds as TLR7 modulators.
  • 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one in the form of a maleate salt will be more easily formulated and/or processed and/or handled.
  • the purity of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one may be improved via formation and/or recrystallisation of a maleate salt, and/or its stability of may be improved vis-à-vis the free base.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one which induces human interferon may be useful in the treatment of various disorders, for example the treatment of allergic diseases and other inflammatory conditions for example allergic rhinitis and asthma, the treatment of infectious diseases and cancer, and may also be useful as a vaccine adjuvant.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one is a potent immunomodulator and accordingly, care should be exercised in its handling.
  • a vaccine adjuvant comprising a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt.
  • an immugenic composition comprising an antigen or antigen composition and a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt.
  • a vaccine composition comprising an antigen or antigen composition and a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt.
  • the invention provides in a further aspect, a combination comprising a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxyl-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, together with at least one other therapeutically-active agent.
  • composition comprising a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, and one or more pharmaceutically acceptable diluents or carriers.
  • a process for preparing a pharmaceutical composition which comprises admixing a compound which is 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, with one or more pharmaceutically acceptable diluents or carriers.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, and salts thereof may be prepared by the methodology described in US Provisional Application Number 61/087777 and International Application No. PCT/EP2009/060265 , published as WO2010/018133 , incorporated herein by reference
  • solvates complexes with solvents in which they are reacted or from which they are precipitated or crystallised.
  • solvents in which they are reacted or from which they are precipitated or crystallised.
  • solvates For example, a complex with water is known as a "hydrate”.
  • Solvents with high boiling points and/or solvents with a high propensity to form hydrogen bonds such as water, ethanol, isopropyl alcohol, and N -methyl pyrrolidinone may be used to form solvates.
  • Methods for the identification of solvates include, but are not limited to, NMR and microanalysis.
  • 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one will be present primarily in the form of the S-isomer, but may include small amounts, for example less than 5%, or less than 3%, and preferably less than 1%, or preferably less than 0.5% of the R-isomer. It will be appreciated that maleate salts of these mixtures are considered within the scope of the present invention.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may exist in tautomeric forms. It will be understood that the present invention encompasses all of the tautomers of 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt, whether as individual tautomers or as mixtures thereof.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one in the form of a maleate salt may be in crystalline or amorphous form. Furthermore, some of the crystalline forms of 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may exist as polymorphs, which are included within the scope of the present invention. The most thermodynamically stable polymorphic form or forms are of particular interest.
  • Polymorphic forms of 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be characterised and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD), infrared spectroscopy (IR), Raman spectroscopy, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid-state nuclear magnetic resonance (ssNMR).
  • XRPD X-ray powder diffraction
  • IR infrared spectroscopy
  • Raman spectroscopy Raman spectroscopy
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • ssNMR solid-state nuclear magnetic resonance
  • 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may have potentially beneficial effects include allergic diseases and other inflammatory conditions for example allergic rhinitis and asthma, infectious diseases, and cancer.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt is also of potential use as a vaccine adjuvant.
  • 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful, as stand-alone or in combination as an adjuvant, in the treatment and/or prevention of immune-mediated disorders, including but not limited to inflammatory or allergic diseases such as asthma, allergic rhinitis and rhinoconjuctivitis, food allergy, hypersensitivity lung diseases, eosinophilic pneumonitis, delayed-type hypersensitivity disorders, atherosclerosis, pancreatitis, gastritis, colitis, osteoarthritis, psoriasis, sarcoidosis, pulmonary fibrosis, respiratory distress syndrome, bronchiolitis, chronic obstructive pulmonary disease, sinusitis, cystic fibrosis, actinic keratosis, skin dysplasia
  • inflammatory or allergic diseases such as
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful in the treatment and/or prevention of reactions against respiratory infections, including but not limited to airways viral exacerbations and tonsillitis.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful in the treatment and/or prevention of autoimmune diseases including but not limited to rheumatoid arthritis, psoriatic arthritis, systemic lupus erythematosus, Sjöegrens disease, ankylosing spondylitis, scleroderma, dermatomyositis, diabetes, graft rejection, including graft-versus-host disease, inflammatory bowel diseases including, but not limited to, Crohn's disease and ulcerative colitis.
  • autoimmune diseases including but not limited to rheumatoid arthritis, psoriatic arthritis, systemic lupus erythematosus, Sjöegrens disease, ankylosing spondylitis, sclero
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful in the treatment of infectious diseases including, but not limited to, those caused by hepatitis viruses (e.g. hepatitis B virus, hepatitis C virus), human immunodeficiency virus, papillomaviruses, herpesviruses, respiratory viruses (e.g. influenza viruses, respiratory syncytial virus, rhinovirus, metapneumovirus, parainfluenzavirus, SARS), and West Nile virus.
  • hepatitis viruses e.g. hepatitis B virus, hepatitis C virus
  • human immunodeficiency virus papillomaviruses
  • herpesviruses papillomaviruses
  • respiratory viruses e.g.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful in the treatment of microbial infections caused by, for example, bacteria, fungi, or protozoa.
  • microbial infections caused by, for example, bacteria, fungi, or protozoa.
  • bacteria fungi, or protozoa
  • these include, but are not limited to, tuberculosis, bacterial pneumonia, aspergillosis, histoplasmosis, candidosis, pneumocystosis, leprosy, chlamydia, cryptococcal disease, cryptosporidosis, toxoplasmosis, leishmania, malaria, and trypanosomiasis.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be useful in the treatment of various cancers, in particular the treatment of cancers that are known to be responsive to immunotherapy and including, but not limited to, renal cell carcinoma, lung cancer, breast cancer, colorectal cancer, bladder cancer, melanoma, leukaemia, lymphomas and ovarian cancer.
  • references herein to treatment or therapy may, depending on the condition, extend to prophylaxis as well as the treatment of established conditions.
  • 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl) pentyl]-7,9-dihydro-8 H -purin-8-one in the form of a maleate salt may be useful as therapeutic agent.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be formulated for administration in any convenient way.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may, for example, be formulated for oral, topical, inhaled, intranasal, buccal, parenteral (for example intravenous, subcutaneous, intradermal, or intramuscular) or rectal administration.
  • 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt is formulated for oral administration.
  • 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt is formulated for topical administration, for example intranasal or inhaled administration.
  • Tablets and capsules for oral administration may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch, cellulose or polyvinyl pyrrolidone; fillers, for example, lactose, microcrystalline cellulose, sugar, maize starch, calcium phosphate or sorbitol; lubricants, for example, magnesium stearate, stearic acid, talc, polyethylene glycol or silica; disintegrants, for example, potato starch, croscarmellose sodium or sodium starch glycollate; or wetting agents such as sodium lauryl sulfate.
  • the tablets may be coated according to methods well known in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, for example, sorbitol syrup, methyl cellulose, glucose/sugar syrup, gelatin, hydroxymethyl cellulose, carboxymethyl cellulose, aluminium stearate gel or hydrogenated edible fats; emulsifying agents, for example, lecithin, sorbitan mono-oleate or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters, propylene glycol or ethyl alcohol; or preservatives, for example, methyl or propyl p-hydroxybenzoates or sorbic acid.
  • the preparations may also contain buffer salts, flavouring, colouring and/or sweetening agents
  • compositions for intranasal administration include aqueous compositions administered to the nose by drops or by pressurised pump. Suitable compositions contain water as the diluent or carrier for this purpose.
  • Compositions for administration to the lung or nose may contain one or more excipients, for example one or more suspending agents, one or more preservatives, one or more surfactants, one or more tonicity adjusting agents, one or more co-solvents, and may include components to control the pH of the composition, for example a buffer system. Further, the compositions may contain other excipients such as antioxidants, for example sodium metabisulfite, and taste-masking agents. Compositions may also be administered to the nose or other regions of the respiratory tract by nebulisation.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may offer sufficient solubility and stability for presentation as an aqueous intranasal solution formulation.
  • Intranasal compositions may permit 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt to be delivered to all areas of the nasal cavities (the target tissue) and further, may permit the active compounds to remain in contact with the target tissue for longer periods of time.
  • a suitable dosing regime for intranasal compositions would be for the patient to inhale slowly through the nose subsequent to the nasal cavity being cleared. During inhalation the composition would be administered to one nostril while the other is manually compressed. This procedure would then be repeated for the other nostril. Typically, one or two sprays per nostril would be administered by the above procedure one, two, or three times each day, ideally once daily. Of particular interest are intranasal compositions suitable for once-daily administration.
  • the suspending agent(s), if included, will typically be present in an amount of from 0.1 to 5% (w/w), such as from 1.5% to 2.4% (w/w), based on the total weight of the composition.
  • pharmaceutically acceptable suspending agents include, but are not limited to, Avicel ® (microcrystalline cellulose and carboxymethylcellulose sodium), carboxymethylcellulose sodium, veegum, tragacanth, bentonite, methylcellulose, xanthan gum, carbopol and polyethylene glycols.
  • compositions for administration to the lung or nose may contain one or more excipients may be protected from microbial or fungal contamination and growth by inclusion of one or more preservatives.
  • pharmaceutically acceptable anti-microbial agents or preservatives include, but are not limited to, quaternary ammonium compounds (for example benzalkonium chloride, benzethonium chloride, cetrimide, cetylpyridinium chloride, lauralkonium chloride and myristyl picolinium chloride), mercurial agents (for example phenylmercuric nitrate, phenylmercuric acetate and thimerosal), alcoholic agents (for example chlorobutanol, phenylethyl alcohol and benzyl alcohol), antibacterial esters (for example esters of para-hydroxybenzoic acid), chelating agents such as disodium edetate (EDTA) and other anti-microbial agents such as chlorhexidine, chlorocresol, sorbic acid and its salts (such
  • Examples of pharmaceutically acceptable anti-fungal agents or preservatives include, but are not limited to, sodium benzoate, sorbic acid, sodium propionate, methylparaben, ethylparaben, propylparaben and butylparaben.
  • the preservative(s), if included, may be present in an amount of from 0.001 to 1% (w/w), such as from 0.015% to 0.5% (w/w) based on the total weight of the composition.
  • compositions may include one or more surfactants which functions to facilitate dissolution of the medicament particles in the aqueous phase of the composition.
  • the amount of surfactant used is an amount which will not cause foaming during mixing.
  • pharmaceutically acceptable surfactants include fatty alcohols, esters and ethers, such as polyoxyethylene (20) sorbitan monooleate (Polysorbate 80), macrogol ethers, and poloxamers.
  • the surfactant may be present in an amount of between about 0.01 to 10% (w/w), such as from 0.01 to 0.75% (w/w), for example about 0.5% (w/w), based on the total weight of the composition.
  • One or more tonicity-adjusting agent(s) may be included to achieve tonicity with body fluids e.g. fluids of the nasal cavity, resulting in reduced levels of irritancy.
  • pharmaceutically acceptable tonicity-adjusting agents include, but are not limited to, sodium chloride, dextrose, xylitol, calcium chloride, glucose, glycerine and sorbitol.
  • a tonicity-adjusting agent, if present, may be included in an amount of from 0.1 to 10% (w/w), such as from 4.5 to 5.5% (w/w), for example about 5.0% (w/w), based on the total weight of the composition.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be buffered by the addition of suitable buffering agents such as sodium citrate, citric acid, trometamol, phosphates such as disodium phosphate (for example the dodecahydrate, heptahydrate, dihydrate and anhydrous forms), or sodium phosphate and mixtures thereof.
  • suitable buffering agents such as sodium citrate, citric acid, trometamol, phosphates such as disodium phosphate (for example the dodecahydrate, heptahydrate, dihydrate and anhydrous forms), or sodium phosphate and mixtures thereof.
  • a buffering agent if present, may be included in an amount of from 0.1 to 5% (w/w), for example 1 to 3% (w/w) based on the total weight of the composition.
  • taste-masking agents include sucralose, sucrose, saccharin or a salt thereof, fructose, dextrose, glycerol, corn syrup, aspartame, acesulfame-K, xylitol, sorbitol, erythritol, ammonium glycyrrhizinate, thaumatin, neotame, mannitol, menthol, eucalyptus oil, camphor, a natural flavouring agent, an artificial flavouring agent, and combinations thereof.
  • co-solvent(s) may be included to aid solubility of the medicament compound(s) and/or other excipients.
  • pharmaceutically acceptable co-solvents include, but are not limited to, propylene glycol, dipropylene glycol, ethylene glycol, glycerol, ethanol, polyethylene glycols (for example PEG300 or PEG400), and methanol.
  • the co-solvent is propylene glycol.
  • Co-solvent(s), if present, may be included in an amount of from 0.05 to 30% (w/w), such as from 1 to 25% (w/w), for example from 1 to 10% (w/w) based on the total weight of the composition.
  • compositions for inhaled administration include aqueous, organic or aqueous/organic mixtures, dry powder or crystalline compositions administered to the respiratory tract by pressurised pump or inhaler, for example, reservoir dry powder inhalers, unit-dose dry powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurised aerosol inhalers, nebulisers or insufflators.
  • Suitable compositions contain water as the diluent or carrier for this purpose and may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like.
  • Aqueous compositions may also be administered to the nose and other regions of the respiratory tract by nebulisation.
  • Such compositions may be aqueous solutions or suspensions or aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • compositions for administration topically to the nose (for example, for the treatment of rhinitis) or to the lung include pressurised aerosol compositions and aqueous compositions delivered to the nasal cavities by pressurised pump.
  • Compositions which are non-pressurised and are suitable for administration topically to the nasal cavity are of particular interest. Suitable compositions contain water as the diluent or carrier for this purpose.
  • Aqueous compositions for administration to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity-modifying agents and the like. Aqueous compositions may also be administered to the nose by nebulisation.
  • a fluid dispenser may typically be used to deliver a fluid composition to the nasal cavities.
  • the fluid composition may be aqueous or non-aqueous, but typically aqueous.
  • Such a fluid dispenser may have a dispensing nozzle or dispensing orifice through which a metered dose of the fluid composition is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser.
  • Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid composition, the doses being dispensable upon sequential pump actuations.
  • the dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid composition into the nasal cavity.
  • a fluid dispenser of the aforementioned type is described and illustrated in International Patent Application publication number WO 2005/044354 (Glaxo Group Limited).
  • the dispenser has a housing which houses a fluid-discharge device having a compression pump mounted on a container for containing a fluid composition.
  • the housing has at least one finger-operable side lever which is movable inwardly with respect to the housing to move the container upwardly in the housing by means of a cam to cause the pump to compress and pump a metered dose of the composition out of a pump stem through a nasal nozzle of the housing.
  • the fluid dispenser is of the general type illustrated in Figures 30-40 of WO 2005/044354 .
  • Aqueous compositions containing 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be delivered by a pump as disclosed in International Patent Application publication number WO2007/138084 (Glaxo Group Limited), for example as disclosed with reference to Figures 22-46 thereof, or as disclosed in United Kingdom patent application number GB0723418.0 (Glaxo Group Limited), for example as disclosed with reference to Figures 7-32 thereof.
  • the pump may be actuated by an actuator as disclosed in Figures 1-6 of GB0723418.0 .
  • Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges of for example gelatine, or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator.
  • Powder blend compositions generally contain a powder mix for inhalation of a maleate salt of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one and a suitable powder base (carrier/diluent/excipient substance) such as mono-, di-, or polysaccharides (for example lactose or starch).
  • Dry powder compositions may also include, in addition to the drug and carrier, a further excipient (for example a ternary agent such as a sugar ester for example cellobiose octaacetate, calcium stearate, or magnesium stearate.
  • a further excipient for example a ternary agent such as a sugar ester for example cellobiose octaacetate, calcium stearate, or magnesium stearate.
  • a composition suitable for inhaled administration may be incorporated into a plurality of sealed dose containers provided on medicament pack(s) mounted inside a suitable inhalation device.
  • the containers may be rupturable, peelable, or otherwise openable one-at-a-time and the doses of the dry powder composition administered by inhalation on a mouthpiece of the inhalation device, as known in the art.
  • the medicament pack may take a number of different forms, for instance a disk-shape or an elongate strip.
  • Representative inhalation devices are the DISKHALERTM and DISKUSTM devices, marketed by GlaxoSmithKline.
  • a dry powder inhalable composition may also be provided as a bulk reservoir in an inhalation device, the device then being provided with a metering mechanism for metering a dose of the composition from the reservoir to an inhalation channel where the metered dose is able to be inhaled by a patient inhaling at a mouthpiece of the device.
  • exemplary marketed devices of this type are TURBUHALERTM (AstraZeneca), TWISTHALERTM (Schering) and CLICKHALERTM (Innovata.)
  • a further delivery method for a dry powder inhalable composition is for metered doses of the composition to be provided in capsules (one dose per capsule) which are then loaded into an inhalation device, typically by the patient on demand.
  • the device has means to rupture, pierce or otherwise open the capsule so that the dose is able to be entrained into the patient's lung when they inhale at the device mouthpiece.
  • ROTAHALERTM GaxoSmithKline
  • HANDIHALERTM Boehringer Ingelheim.
  • Pressurised aerosol compositions suitable for inhalation can be either a suspension or a solution and may contain a maleate salt of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one and a suitable propellant such as a fluorocarbon or hydrogencontaining chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • a suitable propellant such as a fluorocarbon or hydrogencontaining chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • the aerosol composition may optionally contain additional composition excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid or derivative thereof e.g. as described in WO 94/21229 and WO 98/34596 (Minnesota Mining and Manufacturing Company) and co-solvents e.g. ethanol.
  • additional composition excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid or derivative thereof e.g. as described in WO 94/21229 and WO 98/34596 (Minnesota Mining and Manufacturing Company) and co-solvents e.g. ethanol.
  • Pressurised compositions will generally be retained in a canister (e.g. an aluminium canister) closed with a valve (e.g. a metering valve) and fitted into an actuator provided with a mouthpiece.
  • Ointments, creams and gels may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents.
  • bases may thus, for example, include water and/or an oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol.
  • Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, wool-fat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
  • Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch.
  • Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilising agents, suspending agents or preservatives.
  • 6-Amino-2- ⁇ [(1 S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may, for example, be formulated for transdermal delivery by composition into patches or other devices (e.g. pressurised gas devices) which deliver the active component into the skin.
  • devices e.g. pressurised gas devices
  • compositions may take the form of tablets or lozenges formulated in the conventional manner.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be formulated as suppositories, e.g. containing conventional suppository bases such as cocoa butter or other glycerides.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be formulated for parenteral administration by bolus injection or continuous infusion and may be presented in unit dose form, for instance as ampoules, vials, small volume infusions or pre-filled syringes, or in multidose containers with an added preservative.
  • compositions may take such forms as solutions, suspensions, or emulsions in aqueous or non-aqueous vehicles, and may contain formulatory agents such as anti-oxidants, buffers, antimicrobial agents and/or tonicity adjusting agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g. sterile, pyrogen-free water
  • the dry solid presentation may be prepared by filling a sterile powder aseptically into individual sterile containers or by filling a sterile solution aseptically into each container and freeze-drying.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be formulated with vaccines as adjuvants to modulate their activity.
  • compositions may contain antibody(ies) or antibody fragment(s) or an antigenic component including but not limited to protein, DNA, live or dead bacteria and/or viruses or virus-like particles, together with one or more components with adjuvant activity including but not limited to aluminium salts, oil and water emulsions, heat shock proteins, lipid A preparations and derivatives, glycolipids, other TLR agonists such as CpG DNA or similar agents, cytokines such as GM-CSF or IL-12 or similar agents.
  • adjuvant activity including but not limited to aluminium salts, oil and water emulsions, heat shock proteins, lipid A preparations and derivatives, glycolipids, other TLR agonists such as CpG DNA or similar agents, cytokines such as GM-CSF or IL-12 or similar agents.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be employed alone or in combination with other therapeutic agents.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order.
  • the administration of a combination of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one in the form of a maleate salt with other treatment agents may be by administration concomitantly in a unitary pharmaceutical composition including both compounds, or in separate pharmaceutical compositions each including one of the compounds.
  • the combination may be administered separately in a sequential manner wherein one treatment agent is administered first and the other second or vice versa. Such sequential administration may be close in time or remote in time.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be used in combination with one or more agents useful in the prevention or treatment of viral infections.
  • agents include, without limitation; polymerase inhibitors such as those disclosed in WO 2004/037818-A1 , as well as those disclosed in WO 2004/037818 and WO 2006/045613 ; JTK-003, JTK-019, NM-283, HCV-796, R-803, R1728, R1626, as well as those disclosed in WO 2006/018725 , WO 2004/074270 , WO 2003/095441 , US2005/0176701 , WO 2006/020082 , WO 2005/080388 , WO 2004/064925 , WO 2004/065367 , WO 2003/007945 , WO 02/04425 , WO 2005/014543 , WO 2003/000254 , EP 1065213 , WO 01 /47883 , WO 2002/057287 , WO 2002/057245 and similar agents; replication inhibitors such as acyclovir, famciclovir, g
  • 6-Amino-2- ⁇ [(1 S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be used in combination with one or more other agents which may be useful in the prevention or treatment of viral infections for example immune therapies (e.g. interferon or other cytokines/chemokines, cytokine/chemokine receptor modulators, cytokine agonists or antagonists and similar agents); and therapeutic vaccines, antifibrotic agents, anti-inflammatory agents such as corticosteroids or NSAIDs (non-steroidal anti-inflammatory agents) and similar agents.
  • immune therapies e.g. interferon or other cytokines/chemokines, cytokine/chemokine receptor modulators, cytokine agonists or antagonists and similar agents
  • therapeutic vaccines e.g. interferon or other cytokines/chemokines, cytokin
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be used in combination with one or more other agents which may be useful in the prevention or treatment of allergic disease, inflammatory disease, autoimmune disease, for example; antigen immunotherapy, anti-histamines, steroids, NSAIDs, bronchodilators (e.g.
  • beta 2 agonists beta 2 agonists, adrenergic agonists, anticholinergic agents, theophylline), methotrexate, leukotriene modulators and similar agents; monoclonal antibody therapy such as anti-IgE, anti-TNF, anti-IL-5, anti-IL-6, anti-IL-12, anti-IL-1 and similar agents; receptor therapies e.g. entanercept and similar agents; antigen non-specific immunotherapies (e.g. interferon or other cytokines/chemokines, cytokine/chemokine receptor modulators, cytokine agonists or antagonists, TLR agonists and similar agents).
  • monoclonal antibody therapy such as anti-IgE, anti-TNF, anti-IL-5, anti-IL-6, anti-IL-12, anti-IL-1 and similar agents
  • receptor therapies e.g. entanercept and similar agents
  • antigen non-specific immunotherapies e.g. interferon or other cytokines/chemok
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may be used in combination with one or more other agents which may be useful in the prevention or treatment of cancer, for example chemotherapeutics such as alkylating agents, topoisomerase inhibitors, antimetabolites, antimitotic agents, kinase inhibitors and similar agents; monoclonal antibody therapy such as trastuzumab, gemtuzumab and other similar agents; and hormone therapy such as tamoxifen, goserelin and similar agents.
  • chemotherapeutics such as alkylating agents, topoisomerase inhibitors, antimetabolites, antimitotic agents, kinase inhibitors and similar agents
  • monoclonal antibody therapy such as trastuzumab, gemtuzumab and other similar agents
  • hormone therapy such
  • compositions according to the invention may also be used alone or in combination with at least one other therapeutic agent in other therapeutic areas, for example gastrointestinal disease.
  • compositions according to the invention may also be used in combination with gene replacement therapy.
  • the invention includes in a further aspect a combination comprising 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, together with at least one other therapeutically active agent.
  • compositions comprising a combination as defined above together with at least one pharmaceutically acceptable diluent or carrier thereof represent a further aspect of the invention.
  • a therapeutically effective amount of a maleate salt of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one will depend upon a number of factors. For example, the species, age, and weight of the recipient, the precise condition requiring treatment and its severity, the nature of the composition, and the route of administration are all factors to be considered. The therapeutically effective amount ultimately should be at the discretion of the attendant physician.
  • an effective amount of a maleate salt of 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one for the treatment of humans suffering from frailty generally, should be in the range of 0.0001 to 100 mg/kg body weight of recipient per day. More usually the effective amount should be in the range of 0.001 to 10 mg/kg body weight per day. Thus, for a 70 kg adult one example of an actual amount per day would usually be from 7 to 700 mg.
  • typical doses for a 70 kg adult should be in the range of 1 microgramme to 1 mg per day. This amount may be given in a single dose per day or in a number (such as two, three, four, five, or more) of sub-doses per day such that the total daily dose is the same. Similar dosages should be appropriate for treatment of the other conditions referred to herein.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be administered at any appropriate frequency e.g. 1-7 times per week.
  • the precise dosing regimen will of course depend on factors such as the therapeutic indication, the age and condition of the patient, and the particular route of administration chosen.
  • compositions may be presented in unit-dose forms containing a predetermined amount of active ingredient per unit dose.
  • a unit may contain, as a non-limiting example, 0.5 mg to 1 g of a maleate salt of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, depending on the condition being treated, the route of administration, and the age, weight, and condition of the patient.
  • Preferred unit-dosage compositions are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient.
  • Such pharmaceutical compositions may be prepared by any of the methods well-known in the pharmacy art.
  • composition comprising 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, and one or more pharmaceutically acceptable diluents or carriers.
  • the pharmaceutical composition may further comprise at least one other therapeutically active agent.
  • a process for preparing such a pharmaceutical composition which comprises admixing 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt, with one or more pharmaceutically acceptable diluents or carriers.
  • a process for preparing a maleate salt of 6-amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one comprising reacting 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one with a source of maleate anion (for example, maleic acid e.g.
  • the process produces a 1:1 ratio of maleate anion:6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one.
  • 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one, in the form of a maleate salt may be prepared by the methodology described hereinafter.
  • 6-Amino-2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8H-purin-8-one in the form of a maleate salt may also be prepared more generally by reacting 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one with a source of the maleate anion in a suitable solvent e.g isopropyl alcohol.
  • a suitable source of the maleate anion is maleic acid or maleic acid salts.
  • conventional methods of heating and cooling may be employed, for example temperature-regulated oil-baths or temperature-regulated hot-blocks, and ice/salt baths or dry ice/acetone baths respectively.
  • Conventional methods of isolation for example extraction from or into aqueous or non-aqueous solvents may be used.
  • Conventional methods of drying organic solvents, solutions, or extracts such as shaking with anhydrous magnesium sulfate, or anhydrous sodium sulfate, or passing through a hydrophobic frit, may be employed.
  • Conventional methods of purification for example crystallisation and chromatography, for example silica chromatography or reverse-phase chromatography, may be used as required.
  • Crystallisation may be performed using conventional solvents such as ethyl acetate, methanol, ethanol, or butanol, or aqueous mixtures thereof. It will be appreciated that specific reaction times temperatures may typically be determined by reaction-monitoring techniques, for example thin-layer chromatography and LC-MS.
  • the absolute stereochemistry of compounds may be determined using conventional methods, such as X-ray crystallography.
  • Chromatographic purification was typically performed using pre-packed silica gel cartridges.
  • the Flashmaster II is an automated multi-user flash chromatography system, available from Argonaut Technologies Ltd, which utilises disposable, normal phase, Solid Phase Extraction (SPE) cartridges (2 g to 100 g). It provides quaternary on-line solvent mixing to enable gradient methods to be run. Samples are queued using the multi-functional open access software, which manages solvents, flow-rates, gradient profile and collection conditions.
  • the system is equipped with a Knauer variable wavelength UV-detector and two Gilson FC204 fraction-collectors enabling automated peak cutting, collection and tracking.
  • Solvent removal using a stream of nitrogen was performed at 30-40°C on a GreenHouse Blowdown system available from Radleys Discovery Technologies Saffron Walden, Essex, CB11 3AZ, UK
  • UV detection was an averaged signal from wavelength of 210nm to 350nm and mass spectra were recorded on a mass spectrometer using alternate-scan positive and negative mode electrospray ionization.
  • Method A was conducted on an XBridge C 18 column (typically 150mm x 19mm i.d. 5 ⁇ m packing diameter) at ambient temperature.
  • the solvents employed were:
  • 2,6-Dichloro-9-(tetrahydro-2 H -pyran-2-yl)-9 H -purine (36.9 g) (for example, as prepared for Intermediate 1) was heated with 2M ammonia in isopropanol (250 mL) at 50 °C for 5 hours. After standing at ambient temperature overnight, a further quantity of 2M ammonia in isopropanol (100 mL) was added to break up the resultant cake and the reaction mixture was heated for a further 9 hours until the reaction was complete. To the reaction mixture was added water (70 mL) and the yellow solid filtered off.
  • the solid was washed with isopropyl alcohol:water (5:1 (v/v), 60 mL) and then air-dried under suction to give a first crop.
  • the filtrate was re-filtered after standing overnight to isolate precipitate and both solids were dried in vacuo.
  • the first crop was pure with the second crop material showing a very minor impurity (isolated broad signal 3.5 ppm not seen in first crop) but was otherwise identical.
  • Solid first crop 28.4 g
  • solid second crop (3.42 g).
  • N,O-bis(trimethylsilyl)acetamide (975 mL, 3.988 mol) was added to a stirred suspension of 2-fluoro-1 H-purin-6-amine (200 g, 1.306 mmol) (commercially available from, for example, AlliedSignal) in anhydrous acetonitrile (4 L) in a 10L controlled lab reactor and the resulting mixture heated to reflux and maintained at that temperature for 2 hours. The circulator was then re-programmed and the reaction mixture cooled to 0 °C.
  • N-Bromosuccinimide (12.16g, 68.3mmol) was added portionwise over 5 minutes to a stirred solution of 2- ⁇ [(1S)-1-methylbutyl]oxy ⁇ -9-(tetrahydro-2 H -pyran-2-yl)-9H-purin-6-amine (14.9 g, 48.8 mmol) (for example, as prepared for Intermediate 4) in chloroform (80 mL) at ⁇ 5 °C under an atmosphere of nitrogen. The reaction mixture was stirred at ⁇ 5 °C for 5 hours then washed with saturated sodium hydrogen carbonate solution (80 mL) then water (80 mL).
  • the ethyl acetate phase was further diluted with ethyl acetate (4.0 L) and heated to 50 °C. Sulfuric acid (117 mL, 1 eq.) was added dropwise. The reaction mixture was cooled to 10 °C over 1.5 hours and aged for half an hour. The product was isolated by filtration as a white solid, washed on the filter with ethyl acetate (2.4 L) and dried under reduced pressure at 40°C (570 g, 57% theoretical yield).
  • the mixture was stirred for 10 minutes and then allowed to separate with the lower (aqueous) phase retained.
  • the aqueous solution was heated to 50 ⁇ 3 °C for 17 h and then cooled to 20 ⁇ 3 °C.
  • Aqueous sodium hydroxide (2M, ca. 840 mL) was added dropwise until the solution had a pH of 10-11.
  • the resulting suspension was cooled to 10 ⁇ 3 °C, aged for a further 30 min. then filtered.
  • the cake was washed with water (7.6 L) and the product was dried under reduced pressure at 60 °C with a nitrogen bleed to constant weight (207 g, 95%th).
  • X-ray powder diffraction XRPD
  • DSC differential scanning calorimetry
  • XRPD data were acquired on a PANalytical X'Pert Pro powder diffractometer, equipped with an X'Celerator detector.
  • the acquisition conditions were: radiation: Cu K ⁇ , generator tension: 40 kV, generator current: 45 mA, start angle: 2.0° 2 ⁇ , end angle: 40.0° 2 ⁇ , step size: 0.0167° 2 ⁇ .
  • the time per step was 31.750 s.
  • the sample was prepared by mounting a few milligrams of sample on a Si wafer (zero background) plate, resulting in a thin layer of powder.
  • Characteristic peak positions and calculated d-spacings are summarised in Table 1. These were calculated from the raw data using Highscore software. The experimental error in the peak positions is approximately ⁇ 0.1° 2 ⁇ . Relative peak intensities will vary due to preferred orientation.
  • Table 1 Characteristic XRPD Peak Positions for Solid-state Form 1 of 6-Amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one Form 1 2 ⁇ /° d-spacing / ⁇ 5.0 17.6 10.0 8.8 12.7 7.0 13.5 6.5 13.8 6.4 16.6 5.3 18.9 4.7 20.0 4.4 22.2 4.0 23.3 3.8 24.2 3.7 26.1 3.4
  • FIG. 1 A representative XRPD diffractogram of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl) pentyl]-7,9-dihydro-8 H -purin-8-one is shown in Fig. 1 .
  • the DSC thermogram was obtained using a TA Instruments calorimeter. The sample was weighed into an aluminium pan, a pan lid placed on top and lightly crimped without sealing the pan. The experiment was conducted using a heating rate of 10 °C min -1 .
  • FIG. 2 A representative DSC thermogram of 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl) pentyl]-7,9-dihydro-8 H -purin-8-one is shown in Fig. 2 .
  • 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8 H -purin-8-one (for example, as prepared for Reference Example 1) (0.384 g, 0.98 mmol) was dissolved in isopropyl alcohol (4.6 mL, 12 vols) and heated to 40 °C. Maleic acid (0.114 g, 0.98 mmol) was added. A clear solution was obtained. During cooling to room temperature, precipitation occurred. The slurry was filtered, washing with isopropyl alcohol (5 mL) and dried under reduced pressure at 40 °C to constant weight.
  • the resulting slurry was cooled to room temperature and held overnight (approximately 16 hours), then cooled in an ice/water bath for 30 minutes.
  • the slurry was filtered, washing with isopropyl alcohol (4.5 mL, 3 vols and then 3 mL, 2 vols).
  • the product was dried under reduced pressure at 40 °C to constant weight to give 6-amino-2- ⁇ [(1 S )-1-methylbutyl]oxy ⁇ -9-[5-(1-piperidinyl) pentyl]-7,9-dihydro-8 H -purin-8-one, maleate salt (1.305 g, 69%th).
  • the compound of Reference Example 1 was tested for in vitro biological activity in accordance with the following assays, or similar assays:
  • PBMCs Human Peripheral Blood Mononuclear Cells
  • the compound of Reference Example 1 was dissolved in DMSO. Serial 2-fold dilutions with DMSO were prepared and 0.25 ⁇ l dispensed into 384-well clear Greiner polypropylene plates.
  • Blood samples of up to 200 mL were obtained from healthy human donors. Whole blood in 25 mL volumes was overlaid onto 15 mL Ficoll gradients in Leucosep tubes, and centrifuged at 1000 g for 20 min. Cells in the band at the plasma/histopaque interface were carefully removed and washed twice with PBS (centrifuged at 400 g for 5 min to harvest). The final pellet was resuspended in freezing medium (90% Heat-inactivated serum, 10% DMSO) to a cell concentration of 4 ⁇ 10 7 cells/mL. The resuspended cells were then cryopreserved (frozen) using a rate controlled freezer, and stored at -140 °C for up to 4 months.
  • freezing medium 90% Heat-inactivated serum, 10% DMSO
  • PBMCs PBMCs were thawed rapidly in a water bath at 37 °C. A 1:10 dilution of the cells in trypan blue was prepared and counted. The PBMCs were then diluted in growth media [RPMI 1640 containing 10% fetal calf serum (Invitrogen), Penicillin+Streptavidin (Gibco cat.
  • a multi-isoform immunoassay was used to quantify IFN- ⁇ in PBMC supernatants.
  • Rabbit polyclonal antibody against human IFN- ⁇ (catalogue number 31101, Stratech Scientific) was diluted 1:10000 in assay buffer (RPMI 1640 containing 10% fetal calf serum, Invitrogen) and 20 ⁇ L was added to each well of an MSD (Meso-Scale Discovery) single small-spot 384-well GAR (goat anti-rabbit antibody coated) plate. The plate was incubated for 1 hour at room temperature with vigorous shaking. Following three washes with PBS, 20 ⁇ L of cell supernatant were added to each well of the plate. The plate was then incubated for 1 hour at room temperature with vigorous shaking.
  • a pair of monoclonal antibodies to IFN- ⁇ (catalogue numbers 21100 and 21112, Stratech Scientific) were labelled with sulfo-TAG (MSD), diluted 1:1000 in assay buffer and 20 ⁇ L added to each well of the plate. The plate was further incubated for 1 hour at room temperature with vigorous shaking. Following three washes with PBS, 30 ⁇ l of x2 T buffer (MSD) was added to each well and the plate was read on an MSD Sector 6000 plate reader.
  • MSD sulfo-TAG
  • Reference Example 1 had a mean pEC 50 of >8.3
  • PBMCs Mononuclear Cells
  • the compound of Reference Example 1 was dissolved and serially diluted in DMSO to give 100x the required concentration range using a Biomek 2000. 1 ⁇ L of test compound was transferred into 96-well tissue culture plates using a Biomek FX. Each compound was assayed in duplicate for each donor. Each plate contained a dilution series of the TLR7/8 agonist resiquimod as standard and Column 11 contained 1 ⁇ L of 200 ⁇ M resiquimod (giving a 2 ⁇ M final concentration, used to define the approximate maximal response to resiquimod).
  • Blood samples from two human donors were collected into sodium heparin (10 U/mL). 25 mL volumes of whole blood were overlaid onto 15 mL Histopaque in Leucosep tubes which were centrifuged at 800 g for 20 min and the band at the plasma/histopaque interface carefully removed. The collected cells were centrifuged at 2500 rpm for 10 min and the pellet resuspended in 10 mL of media (RPMI 1640 (Low endotoxin) supplemented with 10% v/v foetal calf serum (FCS, low endotoxin) 100 U/mL penicillin G, 100 ⁇ g/mL streptomycin, 10 mM L-glutamine and 1 ⁇ non-essential amino acids).
  • RPMI 1640 Low endotoxin
  • FCS low endotoxin
  • a 1:20 dilution of the cells was prepared using trypan blue and the cells counted using a haemocytometer.
  • the PBMCs were diluted to give a final concentration of 2 ⁇ 10 6 per mL and 100 ⁇ L of this cells suspension was added to wells containing 1 ⁇ L of diluted test compound.
  • the cell preparations were incubated for 24 hr (37 °C, 95% air, 5% CO 2 ) after which a sample of the supernatant was removed using the Biomek FX and assayed for both IFN- ⁇ and TNF- ⁇ using the MSD (Mesoscale Discovery) electrochemiluminescence assay platform.
  • the IFN- ⁇ assay was carried out similarly to that described above.
  • the TNF- ⁇ assay was carried out as per kit instructions (Cat No K111 BHB).
  • Cytokine released was expressed as a percentage of the 2 ⁇ M resiquimod control (column 11). This percentage was plotted against compound concentration and the pEC 50 for the response determined by non-linear least squares curve fitting.
  • a 4 parameter logistic model was selected.
  • TNF responses where a clear maximum response was obtained i.e. a well defined plateau in the response was observed
  • a 4 parameter model was generally used. If the upper asymptote of the curve wasn't well defined then the curve fitting was generally constrained to a maximal response of 100% (i.e. to the response to 2 ⁇ M resiquimod) or to the response of the highest concentration tested if this was greater than the resiquimod response.
  • Reference Example 1 showed a mean pEC 50 for induction of IFN- ⁇ and TNF- ⁇ of ⁇ 9 and ⁇ 6.5 respectively.
  • PBMCs peripheral blood mononuclear cells
  • the compound of Reference Example 1 was dissolved in DMSO, then serially diluted in growth medium (RPMI 1640 medium supplemented with 100 U/mL penicillin G, 100 ⁇ g/mL streptomycin, 10 mM L-glutamine) to give 4x the required concentration range in the presence of 0.04%DMSO. Each compound was assayed in triplicate at all concentrations.
  • Defibrinated human blood from volunteers known to be allergic to Timothy grass was centrifuged at 2500rpm for 15 minutes.
  • the upper layer of serum was collected and heat-inactivated at 56 °C for 30 minutes (HI-autologous serum).
  • the lower layer of cells was resuspended in 50 mL PBS (+Ca +Mg), 25 mL diluted blood were overlaid onto 20 mL Lymphoprep in 50ml tubes then centrifuged at 2500 rpm for 20 minutes at RT.
  • the band at the serum/Lymphoprep interface was carefully removed.
  • the collected cells were washed with PBS and re-suspended at 4 ⁇ 10 6 per mL in growth medium with HI-autologous serum.
  • PBMCs peripheral blood mononuclear cells
  • Reference Example 1 showed mean serum levels of Interferon- ⁇ of 21029 pg/mL. No Interferon- ⁇ was detected in vehicle treated controls.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Otolaryngology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
EP11709025.8A 2010-02-10 2011-02-08 6-amino-2-{[(1s)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8h-purin-8-one maleate Active EP2534149B1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
SI201130364T SI2534149T1 (sl) 2010-02-10 2011-02-08 6-amino-2-((1S)-1-metilbutil)oksi)-9-(5-(1-pipridinil)pentil)-7,9- dihidro-8H purin-8-on maleat
MEP-2014-154A ME01926B (me) 2010-02-10 2011-02-08 6-am in0-2-{[(1s)-1-meti lbuti l]oksi }-9-[5-(1-piperidinil) pentil]-7,9-di hidro-8h- purin-8-0n maleat
PL11709025T PL2534149T3 (pl) 2010-02-10 2011-02-08 Maleinian 6-amino-2-{[(1s)-1-metylobutylo]oksy}-9-[5-(1-piperydynylo)pentylo]-7,9-dihydro-8h-puryn-8-onu
CY20141101069T CY1116091T1 (el) 2010-02-10 2014-12-19 Μηλεϊνικη 6-αμινο-2-{[(1s)-1-μεθυλβουτυλ]οξυ}-9-[5-(1-πιπεριδινυλ)πεντυλ]-7,9-διυδρο-8η-πουριν-8-ονη
HRP20141258AT HRP20141258T1 (hr) 2010-02-10 2014-12-23 6-amino-2-{[(1s)-1-metilbutil]oksi}-9-[5-(1-piperidinil)pentil]-7,9-dihidro-8h-purin-8-on maleat

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30301010P 2010-02-10 2010-02-10
PCT/EP2011/051830 WO2011098452A1 (en) 2010-02-10 2011-02-08 6-amino-2-{ [ (1s)-1-methylbutyl] oxy}-9-[5-(1-piperidinyl)-7,9-dihydro-8h-purin-8-one maleate

Publications (2)

Publication Number Publication Date
EP2534149A1 EP2534149A1 (en) 2012-12-19
EP2534149B1 true EP2534149B1 (en) 2014-10-15

Family

ID=43836817

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11709025.8A Active EP2534149B1 (en) 2010-02-10 2011-02-08 6-amino-2-{[(1s)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8h-purin-8-one maleate

Country Status (23)

Country Link
US (1) US8703754B2 (ru)
EP (1) EP2534149B1 (ru)
JP (1) JP5922587B2 (ru)
KR (1) KR101773541B1 (ru)
CN (1) CN102753553B (ru)
AU (1) AU2011214468B2 (ru)
BR (1) BR112012018904A2 (ru)
CA (1) CA2786973C (ru)
DK (1) DK2534149T3 (ru)
EA (1) EA021048B1 (ru)
ES (1) ES2525826T3 (ru)
HR (1) HRP20141258T1 (ru)
IL (1) IL220845A (ru)
ME (1) ME01926B (ru)
MX (1) MX2012009316A (ru)
PL (1) PL2534149T3 (ru)
PT (1) PT2534149E (ru)
RS (1) RS53745B1 (ru)
SG (1) SG182471A1 (ru)
SI (1) SI2534149T1 (ru)
SM (1) SMT201400196B (ru)
WO (1) WO2011098452A1 (ru)
ZA (1) ZA201205995B (ru)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA103195C2 (ru) 2008-08-11 2013-09-25 Глаксосмитклайн Ллк Производные пурина для применения в лечении аллергий, воспалительных и инфекционных заболеваний
BRPI0923225A2 (pt) 2008-12-02 2016-10-04 Chiralgen Ltd metodo para sintese de acidos nucleicos modificados no atomo de fosforo
KR101885383B1 (ko) 2009-07-06 2018-08-03 웨이브 라이프 사이언시스 리미티드 신규한 핵산 프로드러그 및 그의 사용 방법
NO2491035T3 (ru) 2009-10-22 2018-01-27
JP5868324B2 (ja) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan 不斉補助基
CN103796657B (zh) 2011-07-19 2017-07-11 波涛生命科学有限公司 合成官能化核酸的方法
US10112946B2 (en) * 2011-07-22 2018-10-30 Glaxosmithkline Llc Composition
SG11201500239VA (en) 2012-07-13 2015-03-30 Wave Life Sciences Japan Asymmetric auxiliary group
EP2873674B1 (en) * 2012-07-13 2020-05-06 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
EP4219516A3 (en) 2012-07-13 2024-01-10 Wave Life Sciences Ltd. Chiral control
US9555036B2 (en) 2012-08-24 2017-01-31 Glaxosmithkline Llc Pyrazolopyrimidine compounds
ES2625023T3 (es) 2012-11-20 2017-07-18 Glaxosmithkline Llc Compuestos novedosos
WO2014081644A1 (en) 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
PT2922549T (pt) 2012-11-20 2017-09-01 Glaxosmithkline Llc Novos compostos
WO2015108047A1 (ja) 2014-01-15 2015-07-23 株式会社新日本科学 免疫誘導活性を有するキラル核酸アジュバンド及び免疫誘導活性剤
JPWO2015108048A1 (ja) 2014-01-15 2017-03-23 株式会社新日本科学 抗腫瘍作用を有するキラル核酸アジュバンド及び抗腫瘍剤
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
KR102423317B1 (ko) 2014-01-16 2022-07-22 웨이브 라이프 사이언시스 리미티드 키랄 디자인
EP4218724A3 (en) * 2014-08-29 2023-08-16 Corium Pharma Solutions, Inc. Microstructure array for delivery of active agents
CA2967248A1 (en) * 2014-11-13 2016-05-19 Glaxosmithkline Biologicals Sa Adenine derivatives which are useful in the treatment of allergic diseases or other inflammatory conditions
US20220026404A1 (en) * 2018-11-28 2022-01-27 Shimadzu Corporation Analyzing method for azo compound

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB427857A (en) 1934-08-02 1935-05-01 Newsum Sons & Company Ltd H A new or improved system of construction for skeleton structures, particularly vehicle body frames and door frames
CA2156408C (en) 1993-03-17 2005-02-15 Daniel C. Duan Aerosol formulation containing a polyester dispersing aid
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
CA2310896A1 (en) 1999-07-02 2001-01-02 Japan Tobacco Inc. Hcv polymerase suitable for crystal structure analysis and method for using the enzyme
CA2363274A1 (en) 1999-12-27 2001-07-05 Japan Tobacco Inc. Fused-ring compounds and use thereof as drugs for hepatitis c
US6448281B1 (en) 2000-07-06 2002-09-10 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
US6310224B1 (en) 2001-01-19 2001-10-30 Arco Chemical Technology, L.P. Epoxidation catalyst and process
MY134070A (en) 2001-01-22 2007-11-30 Isis Pharmaceuticals Inc Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
US7285552B2 (en) 2001-03-19 2007-10-23 Ono Pharmaceuticals Co., Ltd. Drugs containing triazaspiro[5.5]undecane derivatives as the active ingredient
AR035543A1 (es) 2001-06-26 2004-06-16 Japan Tobacco Inc Agente terapeutico para la hepatitis c que comprende un compuesto de anillo condensado, compuesto de anillo condensado, composicion farmaceutica que lo comprende, compuestos de benzimidazol, tiazol y bifenilo utiles como intermediarios para producir dichos compuestos, uso del compuesto de anillo con
WO2003007945A1 (en) 2001-07-20 2003-01-30 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
CA2481502A1 (en) 2002-04-04 2003-10-16 Achillion Pharmaceuticals, Inc. Hcv antiviral and cytotoxicity drug screening assay
DOP2003000641A (es) 2002-05-10 2003-11-15 Pfizer Inhibidores de las arn polimerasa dependiente de arn del virus de las hepatitis c y composiciones y tratamiento que los usan
MXPA05004435A (es) 2002-10-24 2005-07-26 Glaxo Group Ltd Derivados de 1-acil-pirrolidina para el tratamiento de infecciones virales.
AU2003300952A1 (en) 2002-12-13 2004-07-09 Smithkline Beecham Corporation Indane compounds as ccr5 antagonists
ES2298627T3 (es) 2002-12-13 2008-05-16 Smithkline Beecham Corporation Compuestos de ciclopropilo como antagonistas de ccr5.
AU2003296992A1 (en) 2002-12-13 2004-07-09 Smithkline Beecham Corporation Pyrrolidine and azetidine compounds as ccr5 antagonists
US7645771B2 (en) 2002-12-13 2010-01-12 Smithkline Beecham Corp. CCR5 antagonists as therapeutic agents
EP1569647B1 (en) 2002-12-13 2008-08-20 Smithkline Beecham Corporation Cyclohexyl compounds as ccr5 antagonists
US7452992B2 (en) 2002-12-13 2008-11-18 Smithkline Beecham Corporation Heterocyclic compounds as CCR5 antagonists
US7098231B2 (en) 2003-01-22 2006-08-29 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7223785B2 (en) 2003-01-22 2007-05-29 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7148226B2 (en) 2003-02-21 2006-12-12 Agouron Pharmaceuticals, Inc. Inhibitors of hepatitis C virus RNA-dependent RNA polymerase, and compositions and treatments using the same
WO2005014543A1 (ja) 2003-08-06 2005-02-17 Japan Tobacco Inc. 縮合環化合物及びそのhcvポリメラーゼ阻害剤としての利用
DK1699512T3 (da) 2003-11-03 2012-09-17 Glaxo Group Ltd Fluiddispenseringsindretning
KR20120091276A (ko) 2004-02-20 2012-08-17 베링거 인겔하임 인터내셔날 게엠베하 바이러스 폴리머라제 억제제
CA2564175A1 (en) 2004-04-28 2005-11-10 Arrow Therapeutics Limited Chemical compounds
US7153848B2 (en) 2004-08-09 2006-12-26 Bristol-Myers Squibb Company Inhibitors of HCV replication
RS51794B (en) 2004-08-18 2011-12-31 Pfizer Inc. RNA SUBSTITUTE RNA POLYMERASE INHIBITORS OF HEPATITIS C VIRUS AND COMPOSITIONS AND TREATMENTS USING THEM
US20060045885A1 (en) 2004-08-27 2006-03-02 Kedl Ross M Method of eliciting an immune response against HIV
GB0423673D0 (en) 2004-10-25 2004-11-24 Glaxo Group Ltd Compounds
SG159561A1 (en) 2005-05-09 2010-03-30 Achillion Pharmaceuticals Inc Thiazole compounds and methods of use
US20090192153A1 (en) * 2005-09-22 2009-07-30 Dainippon Sumitomo Pharma Co., Ltd. a corporation of Japan Novel adenine compound
GB0610666D0 (en) 2006-05-30 2006-07-05 Glaxo Group Ltd Fluid dispenser
CL2008000496A1 (es) 2007-02-19 2008-09-22 Smithkline Beecham Corp Compuestos derivados de purina; procedimiento de preparacion de dichos compuestos; composicion farmaceutica que comprende a dichos compuestos; y su uso para tratar enfermedades infecciosas, cancer, alergias y otras afecciones inflamatorias.
DK2132209T3 (da) * 2007-03-19 2014-04-14 Astrazeneca Ab 9-substituerede 8-oxo-adeninforbindelser som modulatorer af tlr7 (toll-like receptor 7)
PE20091156A1 (es) 2007-12-17 2009-09-03 Astrazeneca Ab Sales de (3-{[[3-(6-amino-2-butoxi-8-oxo-7,8-dihidro-9h-purin-9-il)propil](3-morfolin-4-ilpropil)amino]metil}fenil)acetato de metilo
US8777708B2 (en) 2008-06-27 2014-07-15 Microsoft Corporation Targeting control in a simulated environment
UA103195C2 (ru) 2008-08-11 2013-09-25 Глаксосмитклайн Ллк Производные пурина для применения в лечении аллергий, воспалительных и инфекционных заболеваний

Also Published As

Publication number Publication date
AU2011214468B2 (en) 2015-05-28
SMT201400196B (it) 2015-01-15
RS53745B1 (en) 2015-06-30
EA021048B1 (ru) 2015-03-31
PT2534149E (pt) 2014-12-23
CN102753553A (zh) 2012-10-24
KR101773541B1 (ko) 2017-08-31
PL2534149T3 (pl) 2015-03-31
HRP20141258T1 (hr) 2015-02-27
KR20120135408A (ko) 2012-12-13
SG182471A1 (en) 2012-08-30
AU2011214468A1 (en) 2012-08-09
EA201290631A1 (ru) 2013-03-29
EP2534149A1 (en) 2012-12-19
WO2011098452A1 (en) 2011-08-18
ES2525826T3 (es) 2014-12-30
MX2012009316A (es) 2012-09-12
US20120308609A1 (en) 2012-12-06
ME01926B (me) 2015-05-20
US8703754B2 (en) 2014-04-22
BR112012018904A2 (pt) 2020-09-01
DK2534149T3 (da) 2015-01-05
IL220845A (en) 2016-08-31
ZA201205995B (en) 2014-01-29
CA2786973C (en) 2018-04-10
CA2786973A1 (en) 2011-08-18
CN102753553B (zh) 2016-03-30
JP2013519645A (ja) 2013-05-30
SI2534149T1 (sl) 2015-02-27
JP5922587B2 (ja) 2016-05-24

Similar Documents

Publication Publication Date Title
US10117873B2 (en) 6-amino-purin-8-one compounds
EP2534149B1 (en) 6-amino-2-{[(1s)-1-methylbutyl]oxy}-9-[5-(1-piperidinyl)pentyl]-7,9-dihydro-8h-purin-8-one maleate
US8563717B2 (en) Adenine derivatives
EP2326646B1 (en) Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
US20110135671A1 (en) Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
US8802684B2 (en) Adenine derivatives
WO2010018132A1 (en) Compounds
EP2534148A1 (en) Purine derivatives and their pharmaceutical uses
TWI530289B (zh) 作為人類干擾素誘發物之嘌呤化合物及其組合物和用途

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120809

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

17Q First examination report despatched

Effective date: 20130603

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GLAXOSMITHKLINE LLC

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140521

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 691626

Country of ref document: AT

Kind code of ref document: T

Effective date: 20141115

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011010610

Country of ref document: DE

Effective date: 20141127

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: ISLER AND PEDRAZZINI AG, CH

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20141258

Country of ref document: HR

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20141215

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2525826

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20141230

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20141219

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 5

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E010035

Country of ref document: EE

Effective date: 20150106

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20141258

Country of ref document: HR

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20140402581

Country of ref document: GR

Effective date: 20150128

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20141015

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SK

Ref legal event code: T3

Ref document number: E 17814

Country of ref document: SK

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E022903

Country of ref document: HU

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011010610

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20150716

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 6

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

REG Reference to a national code

Ref country code: NO

Ref legal event code: CHAD

Owner name: GLAXOSMITHKLINE LLC, US

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

REG Reference to a national code

Ref country code: LU

Ref legal event code: HC

Owner name: GLAXOSMITHKLINE LLC; US

Free format text: FORMER OWNER: GLAXOSMITHKLINE LLC

Effective date: 20180110

REG Reference to a national code

Ref country code: FR

Ref legal event code: CA

Effective date: 20180515

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MT

Payment date: 20171211

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20181227

Year of fee payment: 9

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20141258

Country of ref document: HR

Payment date: 20190129

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SM

Payment date: 20181221

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20190206

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CY

Payment date: 20190129

Year of fee payment: 9

Ref country code: CH

Payment date: 20190125

Year of fee payment: 9

Ref country code: FR

Payment date: 20190117

Year of fee payment: 9

Ref country code: NL

Payment date: 20190131

Year of fee payment: 9

Ref country code: RO

Payment date: 20190125

Year of fee payment: 9

Ref country code: CZ

Payment date: 20190124

Year of fee payment: 9

Ref country code: IT

Payment date: 20190213

Year of fee payment: 9

Ref country code: FI

Payment date: 20190129

Year of fee payment: 9

Ref country code: LT

Payment date: 20190129

Year of fee payment: 9

Ref country code: DE

Payment date: 20190115

Year of fee payment: 9

Ref country code: GB

Payment date: 20190128

Year of fee payment: 9

Ref country code: IS

Payment date: 20190115

Year of fee payment: 9

Ref country code: ES

Payment date: 20190301

Year of fee payment: 9

Ref country code: BG

Payment date: 20190124

Year of fee payment: 9

Ref country code: MC

Payment date: 20190130

Year of fee payment: 9

Ref country code: NO

Payment date: 20190129

Year of fee payment: 9

Ref country code: IE

Payment date: 20190129

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20190129

Year of fee payment: 9

Ref country code: HR

Payment date: 20190129

Year of fee payment: 9

Ref country code: RS

Payment date: 20190108

Year of fee payment: 9

Ref country code: SI

Payment date: 20190129

Year of fee payment: 9

Ref country code: SE

Payment date: 20190208

Year of fee payment: 9

Ref country code: EE

Payment date: 20190211

Year of fee payment: 9

Ref country code: LV

Payment date: 20190221

Year of fee payment: 9

Ref country code: BE

Payment date: 20190218

Year of fee payment: 9

Ref country code: TR

Payment date: 20190131

Year of fee payment: 9

Ref country code: DK

Payment date: 20190128

Year of fee payment: 9

Ref country code: AL

Payment date: 20190131

Year of fee payment: 9

Ref country code: AT

Payment date: 20190318

Year of fee payment: 9

Ref country code: HU

Payment date: 20190125

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20190126

Year of fee payment: 9

Ref country code: SK

Payment date: 20190128

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20190130

Year of fee payment: 9

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602011010610

Country of ref document: DE

REG Reference to a national code

Ref country code: HR

Ref legal event code: PBON

Ref document number: P20141258

Country of ref document: HR

Effective date: 20200208

REG Reference to a national code

Ref country code: EE

Ref legal event code: MM4A

Ref document number: E010035

Country of ref document: EE

Effective date: 20200228

REG Reference to a national code

Ref country code: FI

Ref legal event code: MAE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20200229

REG Reference to a national code

Ref country code: NO

Ref legal event code: MMEP

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20200301

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 691626

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200208

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20200208

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20200229

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200909

Ref country code: EE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200302

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200910

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200209

Ref country code: NO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: SM

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200901

REG Reference to a national code

Ref country code: SK

Ref legal event code: MM4A

Ref document number: E 17814

Country of ref document: SK

Effective date: 20200208

REG Reference to a national code

Ref country code: LT

Ref legal event code: MM4D

Effective date: 20200208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: LV

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: SI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200209

Ref country code: BG

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200831

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

Ref country code: RS

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200209

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200301

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200901

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: LT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200229

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20210705

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200209

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200208